1
|
Zheng X, Chen M, Du P, Cairns TC, Zhang L, Gao K, Ni X, Lei Y, Zheng P, Sun J. MstH Is the Major High-Affinity Glucose Transporter with Pleiotropic Roles in Carbon Utilization Regulation and Citric Acid Production in Aspergillus niger. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40350660 DOI: 10.1021/acs.jafc.5c00673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Aspergillus niger is a versatile industrial workhorse for bulk biomanufacturing of organic acids and enzymic proteins owing to its remarkable capacity to utilize diverse crude carbon sources. Glucose, a primary carbon source, plays a critical role in industrial bioprocesses; however, the key components and mechanisms of its dual-affinity transport system in A. niger remain poorly characterized. Herein, we employed transcriptomic profiling and functional analysis of null mutants to comprehensively investigate the glucose transport system. We identified MstH as the principal high-affinity glucose transporter, surpassing MstA in importance, and demonstrated its collaboration with MstC under glucose-abundant conditions. Overexpression of MstH in A. niger significantly enhanced citric acid production by 41.94% compared to that of the parent strain. Furthermore, coexpression of MstH, MstC, and the citrate exporter CexA, along with the deletion of the byproduct-associated genes agdA (α-glucosidase) and oahA (oxaloacetate acetyl hydrolase), led to a citric acid titer of 155.87 ± 0.86 g/L, representing a 3-fold improvement. Transcriptomic analysis revealed that MstH overexpression downregulated the carbon catabolite repressor creA while activating the transcription factor amyR, resulting in upregulation of mstA and mstF and extensive derepression of amylolytic enzymes. Importantly, the point-mutated MstH variant MstHR156 K, with impaired transport function, retained its regulatory effects on creA and amyR, suggesting a moonlighting role for MstH in glucose uptake and carbon utilization regulation. These findings offer new insights into the underlying mechanisms of carbon source utilization and highlight the promising engineering targets for strain optimization in industrial biotechnology.
Collapse
Affiliation(s)
- Xiaomei Zheng
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
- National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meiling Chen
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
- School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Peng Du
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
| | - Timothy C Cairns
- Chair of Applied and Molecular Microbiology, Institute of Biotechnology, Technische Universität Berlin, Berlin 10623, Germany
| | - Lihui Zhang
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
| | - Kaiyue Gao
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
| | - Xiaomeng Ni
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
- National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Yu Lei
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
- National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
| | - Ping Zheng
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
- National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jibin Sun
- Chinese Academy of Sciences, Tianjin Institute of Industrial Biotechnology, Tianjin 300308, China
- National Technology Innovation Center of Synthetic Biology, Tianjin 300308, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
2
|
Guo B, Yu W, Xu X, Liu Y, Liu Y, Du G, Liu L, Lv X. Adaptively Evolved and Multiplexed Engineered Saccharomyces cerevisiae for Neutralizer-Free Production of l-Lactic Acid. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9009-9018. [PMID: 40191959 DOI: 10.1021/acs.jafc.4c12575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
l-Lactic acid is a three-carbon monocarboxylic acid that has extensive applications. However, the bioproduction of l-lactic acid requires the addition of neutralizers, which significantly increases the production costs and can cause environmental pollution. To address this, a Saccharomyces cerevisiae mutant, TMG2, which can tolerate a lactic acid environment (pH 2.60), was obtained through adaptive laboratory evolution. Subsequently, the "push-pull-restrain" strategy was used to improve l-lactic acid production, resulting in a production of 46.8 g/L l-lactic acid. Finally, by overexpressing the transport protein pPfFNT and improving the NADH and acetyl-CoA supply, the l-lactic acid titer of strain TMG27 was improved by 33.8% to 62.6 g/L. Without neutralizers, the l-lactic acid titer reached 76.2 g/L (the fermentation pH was 2.90) with a productivity of 2.1 g/(L h) in a 5-L bioreactor, representing the highest productivity ever reported. Collectively, these results lay the foundation for the environmentally friendly bioproduction of l-lactic acid.
Collapse
Affiliation(s)
- Baoyuan Guo
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Yixing Institute of Food Biotechnology Co., Ltd., Yixing 214200, China
| | - Wenwen Yu
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Xianhao Xu
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yujie Liu
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
- Henan Jindan Lactic Acid Technology Co., Ltd., Dancheng 477100, China
| | - Yanfeng Liu
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Guocheng Du
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Long Liu
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Xueqin Lv
- Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
3
|
Wu N, Wang W, Zhu J. Progress on production of malic acid and succinic acid by industrially-important engineered microorganisms. J Biotechnol 2025; 400:8-19. [PMID: 39923900 DOI: 10.1016/j.jbiotec.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
Organic acids are widely used as additives in the food, pharmaceutical, chemical, and plastic industries. Currently, the industrial production methods of organic acids mainly include plant extraction and chemical synthesis. The latter mainly uses petroleum-based compounds as raw materials to synthesize organic acids through a series of chemical reactions. All of these methods have problems such as environmental pollution, high cost, and unsustainability. By contrast, microbial fermentation can effectively utilize a variety of carbon sources. Due to its low production cost, environmental friendliness, and high product purity, microbial fermentation has received increasing attention in recent years. However, the low yield and long fermentation cycle of microbial fermentation limits its industrial application. With the development of genomics, transcriptomics, and other omics technologies, the metabolic pathways of various strains producing organic acids have gradually been elucidated. Based on this, new technologies such as synthetic biology and high-throughput screening have also been extensively studied. This review summarizes the latest research progress in improving organic acid biosynthesis through metabolic engineering, focusing on L-malic acid (L-MA) and succinic acid (SA). Finally, we also discuss the challenges and future prospects of this field. This review has important reference value in the fields of food, pharmaceuticals, and chemicals, providing a theoretical basis for the study of organic acid biosynthesis.
Collapse
Affiliation(s)
- Na Wu
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, China
| | - Wenxin Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Jianguo Zhu
- Suzhou Wecare Probiotics (Suzhou) Co., Ltd., Suzhou, China.
| |
Collapse
|
4
|
Wang T, Xue H, Liu H, Yuan H, Huang D, Jiang Y. Advancements in metabolic engineering: unlocking the potential of key organic acids for sustainable industrial applications. Front Bioeng Biotechnol 2025; 13:1556516. [PMID: 40134770 PMCID: PMC11933101 DOI: 10.3389/fbioe.2025.1556516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
This review explores the advancements, application potential, and challenges of microbial metabolic engineering strategies for sustainable organic acid production. By integrating gene editing, pathway reconstruction, and dynamic regulation, microbial platforms have achieved enhanced biosynthesis of key organic acids such as pyruvate, lactic acid, and succinic acid. Strategies including by-product pathway knockout, key enzyme overexpression, and improved CO2 fixation have contributed to higher production efficiency. Additionally, utilizing non-food biomass sources, such as lignocellulose, algal feedstocks, and industrial waste, has reduced reliance on conventional carbon sources, supporting sustainability goals. However, challenges remain in substrate inhibition, purification complexity, and metabolic flux imbalances. Addressing these requires omics-driven metabolic optimization, stress-resistant strain development, and biorefinery integration. Future research should focus on system-level design to enhance cost-effectiveness and sustainability, advancing industrial bio-manufacturing of organic acids.
Collapse
Affiliation(s)
- Tengfei Wang
- State Key Laboratory of Green Papermaking and Resource Recycling, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Han Xue
- State Key Laboratory of Green Papermaking and Resource Recycling, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Hongling Liu
- State Key Laboratory of Green Papermaking and Resource Recycling, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Haibo Yuan
- State Key Laboratory of Green Papermaking and Resource Recycling, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Di Huang
- State Key Laboratory of Green Papermaking and Resource Recycling, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| | - Yi Jiang
- State Key Laboratory of Green Papermaking and Resource Recycling, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
- Key Laboratory of Shandong Microbial Engineering, School of Bioengineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China
| |
Collapse
|
5
|
Wu Y, Xu QH, Chen ZL, Yang LH, Guo DS. Synthetic biology meets Aspergillus: engineering strategies for next-generation organic acid production. World J Microbiol Biotechnol 2025; 41:36. [PMID: 39800796 DOI: 10.1007/s11274-024-04246-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 02/27/2025]
Abstract
Organic acids constitute a vital category of chemical raw materials. They have extensive applications in industries such as polymers, food, and pharmaceuticals. Currently, industrial production predominantly relies on microbial fermentation. Aspergillus, due to its unique metabolic capabilities, has become an important microbial resource for organic acid production. In recent years, there has been a growing emphasis on genetic engineering of Aspergillus to increase its yield of organic acids. This review provides a comprehensive overview of the current advancement and future directions in the application of genetic engineering techniques to enhance organic production in Aspergillus, specifically highlighting achievement in reconstructing metabolic pathways for desired products, eliminating by-products, modifying regulatory pathways, and engineering mycelial morphology. Furthermore, this review also focuses on the strategies and genetic tools applied in Aspergillus, with particular emphasis on the potential applications and challenges of CRISPR-based biosensors in organic acid fermentation. By providing insights into these developments, we aim to offer theoretical guidance and innovative approaches for enhancing the efficiency of Aspergillus strains in industrial organic acid production.
Collapse
Affiliation(s)
- Yang Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210023, People's Republic of China
| | - Qian-Hui Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210023, People's Republic of China
| | - Zi-Lei Chen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210023, People's Republic of China
| | - Lin-Hui Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210023, People's Republic of China
| | - Dong-Sheng Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
6
|
Latif A, Hassan N, Ali H, Niazi MBK, Jahan Z, Ghuman IL, Hassan F, Saqib A. An overview of key industrial product citric acid production by Aspergillus niger and its application. J Ind Microbiol Biotechnol 2024; 52:kuaf007. [PMID: 40156584 PMCID: PMC11956825 DOI: 10.1093/jimb/kuaf007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/27/2025] [Indexed: 04/01/2025]
Abstract
Citric acid possesses high economic value and is considered as the world's largest consumed organic acid in numerous industries. Citric acid applications range from food to beverage industries, pharmaceuticals, cosmetics, and the environment. It is mostly produced by microbial fermentation, but Aspergillus niger is considered as the main workhorse for large-scale production of citric acid. In the current review, special devotion has been made toward addressing the latest and innovative literature related to production of citric acid by A. niger. The review article discusses A. niger historical involvement in citric acid production, fermentation technologies, molecular biology, biosynthesis, accumulation of citric acid, methods for enhanced production of citric acid, different operational factors also influencing citric acid production, and various techniques used for citric acid recovery. Also, copious biotechnological applications of citric acid are summarized for a fundamental comprehension of the subject and its critical role in diverse fields of industries. ONE-SENTENCE SUMMARY This review describes the historical role of Aspergillus niger in the production of citric acid, fermentation technologies, molecular biology, techniques for increased citric acid production, and other physical and chemical variables influencing the production of citric acid.
Collapse
Affiliation(s)
- Ambreen Latif
- Industrial Biotechnology Division, National Institute for Biotechnology and Genetics Engineering-College (NIBGE-C), Pakistan Institute of Engineering & Applied Sciences (PIEAS), Faisalabad, Pakistan
- School of Chemical & Materials Engineering, National University for Science and Technology, Islamabad, Pakistan
| | - Noor Hassan
- Industrial Biotechnology Division, National Institute for Biotechnology and Genetics Engineering-College (NIBGE-C), Pakistan Institute of Engineering & Applied Sciences (PIEAS), Faisalabad, Pakistan
| | - Hazrat Ali
- Industrial Biotechnology Division, National Institute for Biotechnology and Genetics Engineering-College (NIBGE-C), Pakistan Institute of Engineering & Applied Sciences (PIEAS), Faisalabad, Pakistan
| | - Muhammad Bilal Khan Niazi
- School of Chemical & Materials Engineering, National University for Science and Technology, Islamabad, Pakistan
- Department of Chemical Engineering, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, Saudi Arabia
- Interdisciplinary Research Center for Refining & Advanced Chemicals, King Fahd University of Petroleum and Minerals (KFUPM), Dhahran, Saudi Arabia
| | - Zaib Jahan
- School of Chemical & Materials Engineering, National University for Science and Technology, Islamabad, Pakistan
| | - Iqra Latif Ghuman
- Industrial Biotechnology Division, National Institute for Biotechnology and Genetics Engineering-College (NIBGE-C), Pakistan Institute of Engineering & Applied Sciences (PIEAS), Faisalabad, Pakistan
| | - Farwa Hassan
- Industrial Biotechnology Division, National Institute for Biotechnology and Genetics Engineering-College (NIBGE-C), Pakistan Institute of Engineering & Applied Sciences (PIEAS), Faisalabad, Pakistan
| | - Anam Saqib
- QuinTech Center for Applied Sciences, Lahore, Pakistan
| |
Collapse
|
7
|
Li B, He J, Zuo K, Xu X, Zou X. Engineering the by-products pathway in Aureobasidium pullulans for highly purified polymalic acid fermentation with concurrent recovery of l-malic acid. BIORESOURCE TECHNOLOGY 2024; 414:131578. [PMID: 39384045 DOI: 10.1016/j.biortech.2024.131578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/05/2024] [Accepted: 10/05/2024] [Indexed: 10/11/2024]
Abstract
The fermentation of polymalic acid (PMA) by Aureobasidium pullulans, followed by acid hydrolysis to release the monomer l-malic acid (l-MA), has emerged as a promising process for the bio-based production of l-MA. However, the presence of specific by-products significantly affects the quality of the final products. In this study, chassis strains harboring an overexpressed endogenous malate dehydrogenase gene (ApMDH2) were engineered to delete key genes involved in the pullulan, melanin, and liamocin biosynthetic pathways. Furthermore, to enhance PMA synthesis productivity and prevent intracellular NADPH accumulation, an irreversible trans-hydrogenase transformation system was designed to efficiently convert NADPH to NADH. In fed-batch fermentation, the engineered strain produced the highest PMA titer (194.3 ± 1.1 g/L) and l-MA yield (0.89 ± 0.01 g/g) with an increased productivity (1.45 ± 0.06 g/L∙h). Moreover, a total of 86.19 % l-MA, with a purity of 99.7 %, was successfully extracted from fermentation broth.
Collapse
Affiliation(s)
- Bingqin Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Jinzhao He
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Kangjia Zuo
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Xingran Xu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China
| | - Xiang Zou
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
8
|
El-Zairy AH, Mohamed HS, Ahmed SA, Ahmed SA, Okla MK, El-Adl K, AbdElgawad H, Hozzein WN. Spectroscopic analysis of wild medicinal desert plants from wadi sanor (beni-suef), Egypt, and their antimicrobial and antioxidant activities. Heliyon 2024; 10:e39612. [PMID: 39553552 PMCID: PMC11564941 DOI: 10.1016/j.heliyon.2024.e39612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/08/2024] [Accepted: 10/18/2024] [Indexed: 11/19/2024] Open
Abstract
Desert plants possess untapped potential for medicinal applications due to their rich phytochemical profiles. However, they need to be more explored. Thus, this study integrates advanced analytical, biochemical, and molecular techniques to investigate the phytochemical composition and biological activities (antimicrobial and antioxidant) of four desert plants (Pergularia tomentosa, Zygophyllum coccineum, Pulicaria undulata, and Ochradenus baccatus), collected from Wadi Sannor, Beni-Suef Governorate, Egypt, in March 2021. The volatile chemicals in the 70 % ethanol extracts of the selected plants were also analyzed using GC-MS. The extract exhibited strong antioxidant properties, as demonstrated by its FRAP (Ferric reducing ability of plasma) values, anti-lipid peroxidation, superoxide anion scavenging activity, and DPPH scavenging activity. Additionally, plants extracts showed high antimicrobial activities against seven pathogens, including three Gram-negative bacteria (Pseudomonas aeruginosa, Salmonella typhimurium, Escherichia coli) and four Gram-positive bacteria (Staphylococcus saprophyticus, Staphylococcus epidermidis, Enterococcus faecalis, Streptococcus salivarius). Lastly, molecular docking was conducted for cis-vaccenic acid, (E)-9-octadecenoic acid, the cyclohepta[b]furan-2-one scaffold, and URS-20(30)-en-3-ol against both the thymidylate kinase enzyme and the active sites of E. coli DNA gyrase. The results from the molecular docking studies showed a strong correlation with the biological data. Moreover, these compounds exhibited good, proposed absorption, distribution, metabolism, and excretion-toxicity (ADMET) profiles. Our study highlights the potential of P. tomentosa, Z. coccineum, P. undulata, and O. baccatus for future medical applications and the development of new pharmaceuticals derived from desert flora.
Collapse
Affiliation(s)
- Amany H. El-Zairy
- Chemistry of medicinal and aromatic plants department, Research Institute of Medicinal and Aromatic plants (RIMAP), Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Hussein S. Mohamed
- Chemistry of medicinal and aromatic plants department, Research Institute of Medicinal and Aromatic plants (RIMAP), Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Shimaa A. Ahmed
- Chemistry department, Faculty of Science, Beni-Suef University, Beni-Suef, 62511, Egypt
| | - Sayed A. Ahmed
- Chemistry department, Faculty of Science, Beni-Suef University, Beni-Suef, 62511, Egypt
- Faculty of Engineering, Nahda University, Beni-Suef, Egypt
| | - Mohammad K. Okla
- Botany and Microbiology Department, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Khaled El-Adl
- Chemistry Department, Faculty of Pharmacy, Heliopolis University for Sustainable Development, Cairo, Egypt
- Pharmaceutical Medicinal Chemistry and Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Hamada AbdElgawad
- Integrated Molecular Plant Physiology Research (IMPRES), Department of Biology, University of Antwerp, Belgium
| | - Wael N. Hozzein
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
9
|
Zhong Y, Shang C, Tao H, Hou J, Cui Z, Qi Q. Boosting succinic acid production of Yarrowia lipolytica at low pH through enhancing product tolerance and glucose metabolism. Microb Cell Fact 2024; 23:291. [PMID: 39443950 PMCID: PMC11515616 DOI: 10.1186/s12934-024-02565-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Succinic acid (SA) is an important bio-based C4 platform chemical with versatile applications, including the production of 1,4-butanediol, tetrahydrofuran, and γ-butyrolactone. The non-conventional yeast Yarrowia lipolytica has garnered substantial interest as a robust cell factory for SA production at low pH. However, the high concentrations of SA, especially under acidic conditions, can impose significant stress on microbial cells, leading to reduced glucose metabolism viability and compromised production performance. Therefore, it is important to develop Y. lipolytica strains with enhanced SA tolerance for industrial-scale SA production. RESULTS An SA-tolerant Y. lipolytica strain E501 with improved SA production was obtained through adaptive laboratory evolution (ALE). In a 5-L bioreactor, the evolved strain E501 produced 89.62 g/L SA, representing a 7.2% increase over the starting strain Hi-SA2. Genome resequencing and transcriptome analysis identified a mutation in the 26S proteasome regulatory subunit Rpn1, as well as genes involved in transmembrane transport, which may be associated with enhanced SA tolerance. By further fine-tuning the glycolytic pathway flux, the highest SA titer of 112.54 g/L to date at low pH was achieved, with a yield of 0.67 g/g glucose and a productivity of 2.08 g/L/h. CONCLUSION This study provided a robust engineered Y. lipolytica strain capable of efficiently producing SA at low pH, thereby reducing the cost of industrial SA fermentation.
Collapse
Affiliation(s)
- Yutao Zhong
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, P. R. China
| | - Changyu Shang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, P. R. China
| | - Huilin Tao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, P. R. China
| | - Jin Hou
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, P. R. China
| | - Zhiyong Cui
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, P. R. China.
| | - Qingsheng Qi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, P. R. China.
| |
Collapse
|
10
|
Wu X, Zhang T, Zhang K, Zhang R, Shi M, Gu C, Shi T, Lu L, Xue F, Xu Q, Zhang C. The forced activation of asexual conidiation in Aspergillus niger simplifies bioproduction. Synth Syst Biotechnol 2024; 9:277-284. [PMID: 38496318 PMCID: PMC10942867 DOI: 10.1016/j.synbio.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/22/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024] Open
Abstract
Aspergillus niger is an efficient cell factory for organic acids production, particularly l-malic acid, through genetic manipulation. However, the traditional method of collecting A. niger spores for inoculation is labor-intensive and resource-consuming. In our study, we used the CRISPR-Cas9 system to replace the promoter of brlA, a key gene in Aspergillus conidiation, with a xylose-inducible promoter xylP in l-malic acid-producing A. niger strain RG0095, generating strain brlAxylP. When induced with xylose in submerged liquid culture, brlAxylP exhibited significant upregulation of conidiation-related genes. This induction allowed us to easily collect an abundance of brlAxylP spores (>7.1 × 106/mL) in liquid xylose medium. Significantly, the submerged conidiation approach preserves the substantial potential of A. niger as a foundational cellular platform for the biosynthesis of organic acids, including but not limited to l-malic acid. In summary, our study offers a simplified submerged conidiation strategy to streamline the preparation stage and reduce labor and material costs for industrial organic acid production using Aspergillus species.
Collapse
Affiliation(s)
- Xingyu Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Tingting Zhang
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Ke Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Rui Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Man Shi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Chenlei Gu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Tianqiong Shi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Ling Lu
- College of Life Sciences, Nanjing Normal University, Nanjing, 210023, China
| | - Feng Xue
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| | - Chi Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, China
| |
Collapse
|
11
|
Zhang C, Shi M, Xu Y, Yang D, Lu L, Xue F, Xu Q. Conditional expression of FumA in Aspergillus niger enhances synthesis of L-malic acid. Appl Environ Microbiol 2024; 90:e0000824. [PMID: 38506527 PMCID: PMC11022578 DOI: 10.1128/aem.00008-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/06/2024] [Indexed: 03/21/2024] Open
Abstract
Currently, the L-malic acid titer achieved through Aspergillus niger fermentation reaches 201 g/L, meeting industrial demands satisfactorily. However, the co-presence of structurally similar fumaric acid and succinic acid in fermentation products suggests a theoretical potential for further improvement in L-malic acid production. In the tricarboxylic acid cycle, fumarate reductase mediates the conversion of succinic acid to fumaric acid. Subsequently, fumarase catalyzes the conversion of fumaric acid to L-malic acid. Notably, both enzymatic reactions are reversible. Our investigation revealed that A. niger contains only one mitochondria-located fumarase FumA. Employing CRISPR-Cas9 technology, we performed a replacement of the fumA promoter with a doxycycline-induced promoter Tet. Under non-inducing condition, the conditional strain exhibited increased levels of fumaric acid and succinic acid. It strongly suggests that FumA mainly promotes the flow of fumaric acid to L-malic acid. Furthermore, a promoter PmfsA that is exclusively activated in a fermentation medium by calcium carbonate was identified through RNA-sequencing screening. Utilizing PmfsA to regulate fumA expression led to a 9.0% increase in L-malic acid titer, an 8.75% increase in yield (glucose to L-malic acid), and an 8.86% enhancement in productivity. This research serves as a significant step toward expediting the industrialization of L-malic acid synthesis via biological fermentation. Additionally, it offers valuable insights for the biosynthesis of other organic acids.IMPORTANCEThis study focuses on enhancing L-malic acid synthesis by modifying the tricarboxylic acid cycle within the mitochondria of Aspergillus niger. We emphasize the significant role of fumarase in converting fumaric acid into L-malic acid, enhancing our understanding of metabolic pathways in A. niger. The precise regulation of fumA is highlighted as a key factor in enhancing L-malic acid production. Furthermore, this research introduces a stringent conditional promoter (PmfsA), exclusively activated by CaCO3. The utilization of PmfsA for fumA expression resulted in heightened L-malic acid titers. The progress in metabolic engineering and bioprocess optimization holds promise for expediting industrial L-malic acid synthesis via biological fermentation. Moreover, it carries implications for the biosynthesis of various other organic acids.
Collapse
Affiliation(s)
- Chi Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Man Shi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Yingyan Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Dongdong Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Ling Lu
- College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Feng Xue
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| |
Collapse
|
12
|
Fan X, Gao X, Zang H, Liu Z, Jing X, Liu X, Guo S, Jiang H, Wu Y, Huang Z, Chen D, Guo R. Transcriptional dynamics and regulatory function of milRNAs in Ascosphaera apis invading Apis mellifera larvae. Front Microbiol 2024; 15:1355035. [PMID: 38650880 PMCID: PMC11033319 DOI: 10.3389/fmicb.2024.1355035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
In the present study, small RNA (sRNA) data from Ascosphaera apis were filtered from sRNA-seq datasets from the gut tissues of A. apis-infected Apis mellifera ligustica worker larvae, which were combined with the previously gained sRNA-seq data from A. apis spores to screen differentially expressed milRNAs (DEmilRNAs), followed by trend analysis and investigation of the DEmilRNAs in relation to significant trends. Additionally, the interactions between the DEmilRNAs and their target mRNAs were verified using a dual-luciferase reporter assay. In total, 974 A. apis milRNAs were identified. The first base of these milRNAs was biased toward U. The expression of six milRNAs was confirmed by stem-loop RT-PCR, and the sequences of milR-3245-y and milR-10285-y were validated using Sanger sequencing. These miRNAs grouped into four significant trends, with the target mRNAs of DEmilRNAs involving 42 GO terms and 120 KEGG pathways, such as the fungal-type cell wall and biosynthesis of secondary metabolites. Further investigation demonstrated that 299 DEmilRNAs (novel-m0011-3p, milR-10048-y, bantam-y, etc.) potentially targeted nine genes encoding secondary metabolite-associated enzymes, while 258 (milR-25-y, milR-14-y, milR-932-x, etc.) and 419 (milR-4561-y, milR-10125-y, let-7-x, etc.) DEmilRNAs putatively targeted virulence factor-encoded genes and nine genes involved in the MAPK signaling pathway, respectively. Additionally, the interaction between ADM-B and milR-6882-x, as well as between PKIA and milR-7009-x were verified. Together, these results not only offer a basis for clarifying the mechanisms underlying DEmilRNA-regulated pathogenesis of A. apis and a novel insight into the interaction between A. apis and honey bee larvae, but also provide candidate DEmilRNA-gene axis for further investigation.
Collapse
Affiliation(s)
- Xiaoxue Fan
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xuze Gao
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
| | - He Zang
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zhitan Liu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xin Jing
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Xiaoyu Liu
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Sijia Guo
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Haibin Jiang
- Jilin Apicultural Research Institute, Jilin, China
| | - Ying Wu
- Jilin Apicultural Research Institute, Jilin, China
| | - Zhijian Huang
- Animal Husbandry Terminus of Sichuan Provincial Department of Agriculture and Rural Affairs, Chengdu, China
| | - Dafu Chen
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
- National and Local United Engineering Laboratory of Natural Biotoxin, Fuzhou, China
- Apitherapy Research Institute of Fujian Province, Fuzhou, China
| | - Rui Guo
- College of Bee Science and Biomedicine, Fujian Agriculture and Forestry University, Fuzhou, China
- National and Local United Engineering Laboratory of Natural Biotoxin, Fuzhou, China
- Apitherapy Research Institute of Fujian Province, Fuzhou, China
| |
Collapse
|
13
|
Yang D, Xu Y, Mo L, Shi M, Wu N, Lu L, Xue F, Xu Q, Zhang C. Enhancing l-Malic Acid Production in Aspergillus niger via Natural Activation of sthA Gene Expression. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:4869-4879. [PMID: 38407053 DOI: 10.1021/acs.jafc.3c09321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The efficient production of l-malic acid using Aspergillus niger requires overcoming challenges in synthesis efficiency and excessive byproduct buildup. This study addresses these hurdles, improving the activity of NADH-dependent malate dehydrogenase (Mdh) in the early stages of the fermentation process. By employing a constitutive promoter to express the Escherichia coli sthA responsible for the transfer of reducing equivalents between NAD(H) and NADP(H) in A. niger, the l-malic acid production was significantly elevated. However, this resulted in conidiation defects of A. niger, limiting industrial viability. To mitigate this, we discovered and utilized the PmfsA promoter, enabling the specific expression of sthA during the fermentation stage. This conditional expression strain showed similar phenotypes to its parent strain while exhibiting exceptional performance in a 5 L fermenter. Notably, it achieved a 65.5% increase in productivity, reduced fermentation cycle by 1.5 days, and lowered succinic acid by 76.2%. This work marks a promising advancement in industrial l-malic acid synthesis via biological fermentation, showcasing the potential of synthetic biology in A. niger for broader applications.
Collapse
Affiliation(s)
- Dongdong Yang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Yingyan Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Li Mo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Man Shi
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Na Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Ling Lu
- College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Feng Xue
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Chi Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
14
|
Xu B, Zhang W, Zhao E, Hong J, Chen X, Wei Z, Li X. Unveiling malic acid biorefinery: Comprehensive insights into feedstocks, microbial strains, and metabolic pathways. BIORESOURCE TECHNOLOGY 2024; 394:130265. [PMID: 38160850 DOI: 10.1016/j.biortech.2023.130265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/24/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
The over-reliance on fossil fuels and resultant environmental issues necessitate sustainable alternatives. Microbial fermentation of biomass for malic acid production offers a viable, eco-friendly solution, enhancing resource efficiency and minimizing ecological damage. This review covers three core aspects of malic acid biorefining: feedstocks, microbial strains, and metabolic pathways. It emphasizes the significance of utilizing biomass sugars, including the co-fermentation of different sugar types to improve feedstock efficiency. The review discusses microbial strains for malic acid fermentation, addressing challenges related to by-products from biomass breakdown and strategies for overcoming them. It delves into the crucial pathways and enzymes for malic acid production, outlining methods to optimize its metabolism, focusing on enzyme regulation, energy balance, and yield enhancement. These insights contribute to advancing the field of consolidated bioprocessing in malic acid biorefining.
Collapse
Affiliation(s)
- Boyang Xu
- Anhui Fermented Food Engineering Research Center, School of Food and Biological Engineering, Hefei University of Technology, Hefei City 230009, Anhui Province, PR China
| | - Wangwei Zhang
- Anhui Fermented Food Engineering Research Center, School of Food and Biological Engineering, Hefei University of Technology, Hefei City 230009, Anhui Province, PR China
| | - Eryong Zhao
- Anhui Fermented Food Engineering Research Center, School of Food and Biological Engineering, Hefei University of Technology, Hefei City 230009, Anhui Province, PR China
| | - Jiong Hong
- School of Life Sciences, University of Science and Technology of China, Hefei City 230026, Anhui Province, PR China
| | - Xiangsong Chen
- Institute of Plasma Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei City 230031, Anhui Province, PR China
| | - Zhaojun Wei
- School of Biological Sciences and Engineering, North Minzu University, Yinchuan City 750030, Ningxia Hui Autonomous Region, PR China.
| | - Xingjiang Li
- Anhui Fermented Food Engineering Research Center, School of Food and Biological Engineering, Hefei University of Technology, Hefei City 230009, Anhui Province, PR China.
| |
Collapse
|
15
|
Wang ZD, Wang BT, Jin L, Ruan HH, Jin FJ. Implications of carbon catabolite repression for Aspergillus-based cell factories: A review. Biotechnol J 2024; 19:e2300551. [PMID: 38403447 DOI: 10.1002/biot.202300551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/19/2023] [Accepted: 12/26/2023] [Indexed: 02/27/2024]
Abstract
Carbon catabolite repression (CCR) is a global regulatory mechanism that allows organisms to preferentially utilize a preferred carbon source (usually glucose) by suppressing the expression of genes associated with the utilization of nonpreferred carbon sources. Aspergillus is a large genus of filamentous fungi, some species of which have been used as microbial cell factories for the production of organic acids, industrial enzymes, pharmaceuticals, and other fermented products due to their safety, substrate convenience, and well-established post-translational modifications. Many recent studies have verified that CCR-related genetic alterations can boost the yield of various carbohydrate-active enzymes (CAZymes), even under CCR conditions. Based on these findings, we emphasize that appropriate regulation of the CCR pathway, especially the expression of the key transcription factor CreA gene, has great potential for further expanding the application of Aspergillus cell factories to develop strains for industrial CAZymes production. Further, the genetically modified CCR strains (chassis hosts) can also be used for the production of other useful natural products and recombinant proteins, among others. We here review the regulatory mechanisms of CCR in Aspergillus and its direct application in enzyme production, as well as its potential application in organic acid and pharmaceutical production to illustrate the effects of CCR on Aspergillus cell factories.
Collapse
Affiliation(s)
- Zhen-Dong Wang
- College of Ecology and Environment, Nanjing Forestry University, Nanjing, China
| | - Bao-Teng Wang
- College of Ecology and Environment, Nanjing Forestry University, Nanjing, China
| | - Long Jin
- College of Ecology and Environment, Nanjing Forestry University, Nanjing, China
| | - Hong-Hua Ruan
- College of Ecology and Environment, Nanjing Forestry University, Nanjing, China
| | - Feng-Jie Jin
- College of Ecology and Environment, Nanjing Forestry University, Nanjing, China
| |
Collapse
|
16
|
Qin Z, Feng J, Li Y, Zheng Y, Moore C, Yang ST. Engineering the reductive tricarboxylic acid pathway in Aureobasidium pullulans for enhanced biosynthesis of poly-L-malic acid. BIORESOURCE TECHNOLOGY 2024; 393:130122. [PMID: 38040309 DOI: 10.1016/j.biortech.2023.130122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/03/2023]
Abstract
Aureobasidium pullulans produced poly-L-malic acid (PMA) as the main metabolite in fermentation but with relatively low productivity and yield limiting its industrial application. In this study, A. pullulans ZX-10 was engineered to overexpress cytosolic malate dehydrogenase (MDH) and pyruvate carboxylase (PYC) and PMA synthetase (PMS) using a high-copy yeast episomal plasmid with the gpdA promoter from Aspergillus nidulans. Overexpressing endogenous PMS and heterologous MDH and PYC from Aspergillus oryzae respectively increased PMA production by 19 % - 37 % (0.64 - 0.74 g/g vs. 0.54 g/g for wild type) in shake-flask fermentations, demonstrating the importance of the reductive tricarboxylic acid (rTCA) pathway in PMA biosynthesis. A. pullulans co-expressing MDH and PYC produced 96.7 g/L PMA at 0.90 g/L∙h and 0.68 g/g glucose in fed-batch fermentation, which were among the highest yield and productivity reported. The engineered A. pullulans with enhanced rTCA pathway is advantageous and promising for PMA production.
Collapse
Affiliation(s)
- Zhen Qin
- William G. Lowrie Department of Chemical & Biomolecular Engineering, The Ohio State University, 151 West Woodruff Ave, Columbus, OH 43210, USA; Department of Food Science and Technology, The Ohio State University, 2015 Fyffe Road, Columbus, OH 43210, USA
| | - Jun Feng
- William G. Lowrie Department of Chemical & Biomolecular Engineering, The Ohio State University, 151 West Woodruff Ave, Columbus, OH 43210, USA
| | - You Li
- William G. Lowrie Department of Chemical & Biomolecular Engineering, The Ohio State University, 151 West Woodruff Ave, Columbus, OH 43210, USA
| | - Yin Zheng
- William G. Lowrie Department of Chemical & Biomolecular Engineering, The Ohio State University, 151 West Woodruff Ave, Columbus, OH 43210, USA
| | - Curtis Moore
- William G. Lowrie Department of Chemical & Biomolecular Engineering, The Ohio State University, 151 West Woodruff Ave, Columbus, OH 43210, USA
| | - Shang-Tian Yang
- William G. Lowrie Department of Chemical & Biomolecular Engineering, The Ohio State University, 151 West Woodruff Ave, Columbus, OH 43210, USA.
| |
Collapse
|
17
|
Zhang K, Zhang TT, Guo RR, Ye Q, Zhao HL, Huang XH. The regulation of key flavor of traditional fermented food by microbial metabolism: A review. Food Chem X 2023; 19:100871. [PMID: 37780239 PMCID: PMC10534219 DOI: 10.1016/j.fochx.2023.100871] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/27/2023] [Accepted: 09/06/2023] [Indexed: 10/03/2023] Open
Abstract
The beneficial microorganisms in food are diverse and complex in structure. These beneficial microorganisms can produce different and unique flavors in the process of food fermentation. The unique flavor of these fermented foods is mainly produced by different raw and auxiliary materials, fermentation technology, and the accumulation of flavor substances by dominant microorganisms during fermentation. The succession and metabolic accumulation of microbial flora significantly impacts the distinctive flavor of fermented foods. The investigation of the role of microbial flora changes in the production of flavor substances during fermentation can reveal the potential connection between microbial flora succession and the formation of key flavor compounds. This paper reviewed the evolution of microbial flora structure as food fermented and the key volatile compounds that contribute to flavor in the food system and their potential relationship. Further, it was a certain guiding significance for food industrial production.
Collapse
Affiliation(s)
- Ke Zhang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Technology Innovation Center for Chinese Prepared Food, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- School of Food and Biological Engineering, Hefei University of Technology, Engineering Research Center of Bio-Process, Ministry of Education, Hefei 230601, Anhui, China
| | - Ting-Ting Zhang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Technology Innovation Center for Chinese Prepared Food, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Ren-Rong Guo
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Technology Innovation Center for Chinese Prepared Food, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Quan Ye
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Technology Innovation Center for Chinese Prepared Food, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Hui-Lin Zhao
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Technology Innovation Center for Chinese Prepared Food, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Xu-Hui Huang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Dalian Technology Innovation Center for Chinese Prepared Food, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
18
|
Lv X, Jin K, Yi Y, Song L, Xiu X, Liu Y, Li J, Du G, Chen J, Liu L. Analysis of acid-tolerance mechanism based on membrane microdomains in Saccharomyces cerevisiae. Microb Cell Fact 2023; 22:180. [PMID: 37700284 PMCID: PMC10498586 DOI: 10.1186/s12934-023-02195-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/05/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Saccharomyces cerevisiae has been used in the biosynthesis of acid products such as organic acids owing to its acid tolerance. Improving the acid tolerance of S. cerevisiae is beneficial for expanding its application range. Our previous study isolated the TAMC strain that was tolerant to a pH 2.3 through adaptive laboratory evolution; however, its mechanism underlying tolerance to low pH environment remains unclear. RESULTS In this study, through visual observation and order analysis of plasma membrane and membrane microdomains, we revealed that the membrane microdomains of TAMC strain play an indispensable role in acid tolerance. Transcriptomic analysis showed an increase in the expression of genes related to key components of membrane microdomains in TAMC strain. Furthermore, an obvious reduction was observed in the acid tolerance of the strain with sterol C-24 methyltransferase encoding gene ERG6 knockout for inhibiting membrane microdomain formation. Finally, colocalization analysis of H+-ATPase PMA1 and plasma membrane protein PMP1 showed that disruption of membrane microdomains could inhibit the formation of the H+-ATPase complex. CONCLUSIONS Membrane microdomains could provide a platform for forming H+-ATPase complexes to facilitate intracellular H+ homeostasis, and thereby improve cell acid resistance. This study proposed a novel acid tolerance mechanism, providing a new direction for the rational engineering of acid-tolerant strains.
Collapse
Affiliation(s)
- Xueqin Lv
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Yixing Institute of Food Biotechnology Co., Ltd, Yixing, 214200, China
| | - Ke Jin
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Yixing Institute of Food Biotechnology Co., Ltd, Yixing, 214200, China
| | - Yu Yi
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
| | - Lingang Song
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
| | - Xiang Xiu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
| | - Yanfeng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
| | - Jianghua Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
| | - Guocheng Du
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
| | - Jian Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi, 214122, China
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China
- Yixing Institute of Food Biotechnology Co., Ltd, Yixing, 214200, China
| | - Long Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi, 214122, China.
- Science Center for Future Foods, Jiangnan University, Wuxi, 214122, China.
- Yixing Institute of Food Biotechnology Co., Ltd, Yixing, 214200, China.
- Food Laboratory of Zhongyuan, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
19
|
Zhou S, Ding N, Han R, Deng Y. Metabolic engineering and fermentation optimization strategies for producing organic acids of the tricarboxylic acid cycle by microbial cell factories. BIORESOURCE TECHNOLOGY 2023; 379:128986. [PMID: 37001700 DOI: 10.1016/j.biortech.2023.128986] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 05/03/2023]
Abstract
The organic acids of the tricarboxylic acid (TCA) pathway are important platform compounds and are widely used in many areas. The high-productivity strains and high-efficient and low-cost fermentation are required to satisfy a huge market size. The high metabolic flux of the TCA pathway endows microorganisms potential to produce high titers of these organic acids. Coupled with metabolic engineering and fermentation optimization, the titer of the organic acids has been significantly improved in recent years. Herein, we discuss and compare the recent advances in synthetic pathway engineering, cofactor engineering, transporter engineering, and fermentation optimization strategies to maximize the biosynthesis of organic acids. Such engineering strategies were mainly based on the TCA pathway and glyoxylate pathway. Furthermore, organic-acid-secretion enhancement and renewable-substrate-based fermentation are often performed to assist the biosynthesis of organic acids. Further strategies are also discussed to construct high-productivity and acid-resistant strains for industrial large-scale production.
Collapse
Affiliation(s)
- Shenghu Zhou
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Nana Ding
- College of Food and Health, Zhejiang A&F University, Hangzhou 311300, China
| | - Runhua Han
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, United States
| | - Yu Deng
- National Engineering Research Center for Cereal Fermentation and Food Biomanufacturing, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China; Jiangsu Provincial Research Center for Bioactive Product Processing Technology, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China.
| |
Collapse
|
20
|
Chen Z, Zhang C, Pei L, Qian Q, Lu L. Production of L-Malic Acid by Metabolically Engineered Aspergillus nidulans Based on Efficient CRISPR-Cas9 and Cre- loxP Systems. J Fungi (Basel) 2023; 9:719. [PMID: 37504708 PMCID: PMC10381526 DOI: 10.3390/jof9070719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
Aspergillus nidulans has been more extensively characterized than other Aspergillus species considering its morphology, physiology, metabolic pathways, and genetic regulation. As it has a rapid growth rate accompanied by simple nutritional requirements and a high tolerance to extreme cultural conditions, A. nidulans is a promising microbial cell factory to biosynthesize various products in industry. However, it remains unclear for whether it is also a suitable host for synthesizing abundant L-malic acid. In this study, we developed a convenient and efficient double-gene-editing system in A. nidulans strain TN02A7 based on the CRISPR-Cas9 and Cre-loxP systems. Using this gene-editing system, we made a L-malic acid-producing strain, ZQ07, derived from TN02A7, by deleting or overexpressing five genes (encoding Pyc, pyruvate carboxylase; OahA, oxaloacetate acetylhydrolase; MdhC, malate dehydrogenase; DctA, C4-dicarboxylic acid transporter; and CexA, citric acid transporter). The L-malic acid yield in ZQ07 increased to approximately 9.6 times higher (up to 30.7 g/L titer) than that of the original unedited strain TN02A7, in which the production of L-malic acid was originally very low. The findings in this study not only demonstrate that A. nidulans could be used as a potential host for biosynthesizing organic acids, but also provide a highly efficient gene-editing strategy in filamentous fungi.
Collapse
Affiliation(s)
- Ziqing Chen
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Chi Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Lingling Pei
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Qi Qian
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Ling Lu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, Jiangsu Engineering and Technology Research Centre for Microbiology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
21
|
Khandelwal R, Srivastava P, Bisaria VS. Recent advances in the production of malic acid by native fungi and engineered microbes. World J Microbiol Biotechnol 2023; 39:217. [PMID: 37269376 DOI: 10.1007/s11274-023-03666-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/25/2023] [Indexed: 06/05/2023]
Abstract
Malic acid is mainly produced by chemical methods which lead to various environmental sustainability concerns associated with CO2 emissions and resulting global warming. Since malic acid is naturally synthesized, microorganisms offer an eco-friendly and cost-effective alternative for its production. An additional advantage of microbial production is the synthesis of pure L-form of malic acid. Due to its numerous applications, biotechnologically- produced L-malic acid is a much sought-after platform chemical. Malic acid can be produced by microbial fermentation via oxidative/reductive TCA and glyoxylate pathways. This article elaborates the potential and limitations of high malic acid producing native fungi belonging to Aspergillus, Penicillium, Ustilago and Aureobasidium spp. The utilization of industrial side streams and low value renewable substrates such as crude glycerol and lignocellulosic biomass is also discussed with a view to develop a competitive bio-based production process. The major impediments present in the form of toxic compounds from lignocellulosic residues or synthesized during fermentation along with their remedial measures are also described. The article also focuses on production of polymalic acid from renewable substrates which opens up a cost-cutting dimension in production of this biodegradable polymer. Finally, the recent strategies being employed for its production in recombinant organisms have also been covered.
Collapse
Affiliation(s)
- Rohit Khandelwal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
- Corporate Research & Development Centre, Bharat Petroleum Corporation Limited, Udyog Kendra, P. O. Surajpur, Greater Noida, 201306, India
| | - Preeti Srivastava
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Virendra Swarup Bisaria
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India.
| |
Collapse
|
22
|
Zuo H, Ji L, Pan J, Chen X, Gao C, Liu J, Wei W, Wu J, Song W, Liu L. Engineering growth phenotypes of Aspergillus oryzae for L-malate production. BIORESOUR BIOPROCESS 2023; 10:25. [PMID: 38647943 PMCID: PMC10991988 DOI: 10.1186/s40643-023-00642-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/09/2023] [Indexed: 04/09/2023] Open
Abstract
Improving the growth status of Aspergillus oryzae is an efficient way to enhance L-malate production. However, the growth mechanism of filamentous fungi is relatively complex, which limits A. oryzae as a cell factory to produce L-malate industrially. This study determined the relationship between growth status and L-malate production. The optimal ranges of colony diameter, percentage of vegetative mycelia, and pellet number of A. oryzae were determined to be 26-30 mm, 35-40%, and 220-240/mL, respectively. To achieve this optimum range, adaptive evolution was used to obtain the evolved strain Z07 with 132.54 g/L L-malate and a productivity of 1.1 g/L/h. Finally, a combination of transcriptome analysis and morphological characterization was used to identify the relevant pathway genes that affect the growth mechanism of A. oryzae. The strategies used in this study and the growth mechanism provide a good basis for efficient L-malate production by filamentous fungi.
Collapse
Affiliation(s)
- Huiyun Zuo
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Lihao Ji
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Jingyu Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Xiulai Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Cong Gao
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Jia Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Wanqing Wei
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Jing Wu
- School of Pharmaceutical Science, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Wei Song
- School of Pharmaceutical Science, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Liming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi, 214122, Jiangsu, China.
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
23
|
Ding Q, Ye C. Recent advances in producing food additive L-malate: Chassis, substrate, pathway, fermentation regulation and application. Microb Biotechnol 2023; 16:709-725. [PMID: 36604311 PMCID: PMC10034640 DOI: 10.1111/1751-7915.14206] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/22/2022] [Indexed: 01/07/2023] Open
Abstract
In addition to being an important intermediate in the TCA cycle, L-malate is also widely used in the chemical and beverage industries. Due to the resulting high demand, numerous studies investigated chemical methods to synthesize L-malate from petrochemical resources, but such approaches are hampered by complex downstream processing and environmental pollution. Accordingly, there is an urgent need to develop microbial methods for environmentally-friendly and economical L-malate biosynthesis. The rapid progress and understanding of DNA manipulation, cell physiology, and cell metabolism can improve industrial L-malate biosynthesis by applying intelligent biochemical strategies and advanced synthetic biology tools. In this paper, we mainly focused on biotechnological approaches for enhancing L-malate synthesis, encompassing the microbial chassis, substrate utilization, synthesis pathway, fermentation regulation, and industrial application. This review emphasizes the application of novel metabolic engineering strategies and synthetic biology tools combined with a deep understanding of microbial physiology to improve industrial L-malate biosynthesis in the future.
Collapse
Affiliation(s)
- Qiang Ding
- School of Life SciencesAnhui UniversityHefeiChina
- Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education InstitutesAnhui UniversityHefeiChina
- Anhui Key Laboratory of Modern BiomanufacturingHefeiChina
| | - Chao Ye
- School of Food Science and Pharmaceutical EngineeringNanjing Normal UniversityNanjingChina
| |
Collapse
|
24
|
Chen Y, Han A, Wang M, Wei D, Wang W. Metabolic Engineering of Trichoderma reesei for l-Malic Acid Production. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4043-4050. [PMID: 36812909 DOI: 10.1021/acs.jafc.2c09078] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
l-Malic acid has various applications in the chemical and food industries. The filamentous fungus Trichoderma reesei is known to be an efficient enzyme producer. Here, through metabolic engineering, T. reesei was constructed for the first time as an excellent cell factory for l-malic acid production. The heterologous overexpression of genes encoding the C4-dicarboxylate transporter from Aspergillus oryzae and Schizosaccharomyces pombe initiated l-malic acid production. The overexpression of pyruvate carboxylase from A. oryzae in the reductive tricarboxylic acid pathway further increased both the titer and yield of l-malic acid, resulting in the highest titer reported in a shake-flask culture. Furthermore, the deletion of malate thiokinase blocked l-malic acid degradation. Finally, the engineered T. reesei strain produced 220.5 g/L of l-malic acid in a 5 L fed-batch culture (productivity of 1.15 g/L/h). A T. reesei cell factory was created for the efficient production of l-malic acid.
Collapse
Affiliation(s)
- Yumeng Chen
- State Key Lab of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ao Han
- State Key Lab of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Meng Wang
- State Key Lab of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Dongzhi Wei
- State Key Lab of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Wei Wang
- State Key Lab of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
25
|
Effective production of kojic acid in engineered Aspergillus niger. Microb Cell Fact 2023; 22:40. [PMID: 36843006 PMCID: PMC9969635 DOI: 10.1186/s12934-023-02038-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/08/2023] [Indexed: 02/28/2023] Open
Abstract
BACKGROUND Kojic acid (KA) is a widely used compound in the cosmetic, medical, and food industries, and is typically produced by Aspergillus oryzae. To meet increasing market demand, it is important to optimize KA production through seeking alternatives that are more economic than current A. oryzae-based methods. RESULTS In this study, we achieved the first successful heterologous production of KA in Aspergillus niger, an industrially important fungus that does not naturally produce KA, through the expression of the kojA gene from A. oryzae. Using the resulting KA-producing A. niger strain as a platform, we identified four genes (nrkA, nrkB, nrkC, and nrkD) that negatively regulate KA production. Knocking down nrkA or deleting any of the other three genes resulted in a significant increase in KA production in shaking flask cultivation. The highest KA titer (25.71 g/L) was achieved in a pH controlled batch bioreactor using the kojA overexpression strain with a deletion of nrkC, which showed a 26.7% improvement compared to the KA titer (20.29 g/L) that was achieved in shaking flask cultivation. CONCLUSION Our study demonstrates the potential of using A. niger as a platform for studying KA biosynthesis and regulation, and for the cost-effective production of KA in industrial strain development.
Collapse
|
26
|
Ploessl D, Zhao Y, Shao Z. Engineering of non-model eukaryotes for bioenergy and biochemical production. Curr Opin Biotechnol 2023; 79:102869. [PMID: 36584447 DOI: 10.1016/j.copbio.2022.102869] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/14/2022] [Accepted: 11/23/2022] [Indexed: 12/29/2022]
Abstract
The prospect of leveraging naturally occurring phenotypes to overcome bottlenecks constraining the bioeconomy has marshalled increased exploration of nonconventional organisms. This review discusses the status of non-model eukaryotic species in bioproduction, the evaluation criteria for effectively matching a candidate host to a biosynthetic process, and the genetic engineering tools needed for host domestication. We present breakthroughs in genome editing and heterologous pathway design, delving into innovative spatiotemporal modulation strategies that potentiate more refined engineering capabilities. We cover current understanding of genetic instability and its ramifications for industrial scale-up, highlighting key factors and possible remedies. Finally, we propose future opportunities to expand the current collection of available hosts and provide guidance to benefit the broader bioeconomy.
Collapse
Affiliation(s)
- Deon Ploessl
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; NSF Engineering Research Center for Biorenewable Chemicals, Iowa State University, Ames, IA, USA
| | - Yuxin Zhao
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; NSF Engineering Research Center for Biorenewable Chemicals, Iowa State University, Ames, IA, USA; DOE Center for Advanced Bioenergy and Bioproducts Innovation, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Zengyi Shao
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, USA; NSF Engineering Research Center for Biorenewable Chemicals, Iowa State University, Ames, IA, USA; DOE Center for Advanced Bioenergy and Bioproducts Innovation, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Interdepartmental Microbiology Program, Iowa State University, Ames, IA, USA; Bioeconomy Institute, Iowa State University, Ames, IA, USA; The Ames Laboratory, Ames, IA, USA.
| |
Collapse
|
27
|
Sun L, Zhang Q, Kong X, Liu Y, Li J, Du G, Lv X, Ledesma-Amaro R, Chen J, Liu L. Highly efficient neutralizer-free l-malic acid production using engineered Saccharomyces cerevisiae. BIORESOURCE TECHNOLOGY 2023; 370:128580. [PMID: 36608859 DOI: 10.1016/j.biortech.2023.128580] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
In industrial bioproduction of organic acids, numerous neutralizers are required which substantially increases production costs and burdens the environment. To address this challenge, a Saccharomyces cerevisiae mutant (named TAMC) with a low pH tolerance (pH 2.3) was isolated by adaptive laboratory evolution. Taking the synthesis of l-malic acid as an example, the malate dehydrogenase 3 without signal peptide (MDHΔSKL) and pyruvate carboxylase 2 (PYC2) were overexpressed in cytoplasmic synthesis pathway, and the l-malic acid titer increased 5.6-fold. Subsequently, the malic acid transporter SpMae1 was designed, and the extracellular l-malic acid titer was increased from 7.3 to 73.6 g/L. Furthermore, by optimizing the synthesis of the precursor pyruvate, the titer reached 81.8 g/L. Finally, without any neutralizer, the titer in the 3-L bioreactor reached 232.9 g/L, the highest l-malic acid titer reported to date. Herein, the engineered l-malic acid overproducer paves the way for the large-scale green production of l-malic acid.
Collapse
Affiliation(s)
- Li Sun
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Quanwei Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Xiao Kong
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Yanfeng Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Jianghua Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Guocheng Du
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Xueqin Lv
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Rodrigo Ledesma-Amaro
- Department of Bioengineering and Centre for Synthetic Biology, Imperial College London, London SW7 2AZ, UK
| | - Jian Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China
| | - Long Liu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, Jiangnan University, Wuxi 214122, China; Science Center for Future Foods, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
28
|
Ding Q, Ye C. Microbial cell factories based on filamentous bacteria, yeasts, and fungi. Microb Cell Fact 2023; 22:20. [PMID: 36717860 PMCID: PMC9885587 DOI: 10.1186/s12934-023-02025-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 01/20/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Advanced DNA synthesis, biosensor assembly, and genetic circuit development in synthetic biology and metabolic engineering have reinforced the application of filamentous bacteria, yeasts, and fungi as promising chassis cells for chemical production, but their industrial application remains a major challenge that needs to be solved. RESULTS As important chassis strains, filamentous microorganisms can synthesize important enzymes, chemicals, and niche pharmaceutical products through microbial fermentation. With the aid of metabolic engineering and synthetic biology, filamentous bacteria, yeasts, and fungi can be developed into efficient microbial cell factories through genome engineering, pathway engineering, tolerance engineering, and microbial engineering. Mutant screening and metabolic engineering can be used in filamentous bacteria, filamentous yeasts (Candida glabrata, Candida utilis), and filamentous fungi (Aspergillus sp., Rhizopus sp.) to greatly increase their capacity for chemical production. This review highlights the potential of using biotechnology to further develop filamentous bacteria, yeasts, and fungi as alternative chassis strains. CONCLUSIONS In this review, we recapitulate the recent progress in the application of filamentous bacteria, yeasts, and fungi as microbial cell factories. Furthermore, emphasis on metabolic engineering strategies involved in cellular tolerance, metabolic engineering, and screening are discussed. Finally, we offer an outlook on advanced techniques for the engineering of filamentous bacteria, yeasts, and fungi.
Collapse
Affiliation(s)
- Qiang Ding
- grid.252245.60000 0001 0085 4987School of Life Sciences, Anhui University, Hefei, 230601 China ,grid.252245.60000 0001 0085 4987Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, 230601 Anhui China ,Anhui Key Laboratory of Modern Biomanufacturing, Hefei, 230601 Anhui China
| | - Chao Ye
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China
| |
Collapse
|
29
|
Xi Y, Xu H, Zhan T, Qin Y, Fan F, Zhang X. Metabolic engineering of the acid-tolerant yeast Pichia kudriavzevii for efficient L-malic acid production at low pH. Metab Eng 2023; 75:170-180. [PMID: 36566973 DOI: 10.1016/j.ymben.2022.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/27/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Currently, the biological production of L-malic acid (L-MA) is mainly based on the fermentation of filamentous fungi at near-neutral pH, but this process requires large amounts of neutralizing agents, resulting in the generation of waste salts when free acid is obtained in the downstream process, and the environmental hazards associated with the waste salts limit the practical application of this process. To produce L-MA in a more environmentally friendly way, we metabolically engineered the acid-tolerant yeast Pichia kudriavzevii and achieved efficient production of L-MA through low pH fermentation. First, an initial L-MA-producing strain that relies on the reductive tricarboxylic acid (rTCA) pathway was constructed. Subsequently, the L-MA titer and yield were further increased by fine-tuning the flux between the pyruvate and oxaloacetate nodes. In addition, we found that the insufficient supply of NADH for cytoplasmic malate dehydrogenase (MDH) hindered the L-MA production at low pH, which was resolved by overexpressing the soluble pyridine nucleotide transhydrogenase SthA from E. coli. Transcriptomic and metabolomic data showed that overexpression of EcSthA contributed to the activation of the pentose phosphate pathway and provided additional reducing power for MDH by converting NADPH to NADH. Furthermore, overexpression of EcSthA was found to help reduce the accumulation of the by-product pyruvate but had no effect on the accumulation of succinate. In microaerobic batch fermentation in a 5-L fermenter, the best strain, MA009-10-URA3 produced 199.4 g/L L-MA with a yield of 0.94 g/g glucose (1.27 mol/mol), with a productivity of 1.86 g/L/h. The final pH of the fermentation broth was approximately 3.10, meaning that the amount of neutralizer used was reduced by more than 50% compared to the common fermentation processes using filamentous fungi. To our knowledge, this is the first report of the efficient bioproduction of L-MA at low pH and represents the highest yield of L-MA in yeasts reported to date.
Collapse
Affiliation(s)
- Yongyan Xi
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, 32 West 7th Avenue, Tianjin Airport Economic Area, Tianjin, 300308, PR China; Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China; National Innovation Center for Synthetic Biotechnology, Tianjin, 300308, PR China
| | - Hongtao Xu
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, 32 West 7th Avenue, Tianjin Airport Economic Area, Tianjin, 300308, PR China; Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China; National Innovation Center for Synthetic Biotechnology, Tianjin, 300308, PR China
| | - Tao Zhan
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, 32 West 7th Avenue, Tianjin Airport Economic Area, Tianjin, 300308, PR China; Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China; National Innovation Center for Synthetic Biotechnology, Tianjin, 300308, PR China
| | - Ying Qin
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, 32 West 7th Avenue, Tianjin Airport Economic Area, Tianjin, 300308, PR China; Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China; National Innovation Center for Synthetic Biotechnology, Tianjin, 300308, PR China
| | - Feiyu Fan
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, 32 West 7th Avenue, Tianjin Airport Economic Area, Tianjin, 300308, PR China; Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China; National Innovation Center for Synthetic Biotechnology, Tianjin, 300308, PR China.
| | - Xueli Zhang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, 32 West 7th Avenue, Tianjin Airport Economic Area, Tianjin, 300308, PR China; Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, PR China; National Innovation Center for Synthetic Biotechnology, Tianjin, 300308, PR China.
| |
Collapse
|
30
|
Wu N, Xing M, Chen Y, Zhang C, Li Y, Song P, Xu Q, Liu H, Huang H. Improving the productivity of malic acid by alleviating oxidative stress during Aspergillus niger fermentation. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2022; 15:151. [PMID: 36581946 PMCID: PMC9801644 DOI: 10.1186/s13068-022-02250-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/15/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND As an attractive platform chemical, malic acid has been commonly used in the food, feed and pharmaceutical field. Microbial fermentation of biobased sources to produce malic acid has attracted great attention because it is sustainable and environment-friendly. However, most studies mainly focus on improving yield and ignore shortening fermentation time. A long fermentation period means high cost, and hinders the industrial applications of microbial fermentation. Stresses, especially oxidative stress generated during fermentation, inhibit microbial growth and production, and prolong fermentation period. Previous studies have shown that polypeptides could effectively relieve stresses, but the underlying mechanisms were poorly understood. RESULTS In this study, polypeptides (especially elastin peptide) addition improves the productivity of malic acid in A. niger, resulting in shortening of fermentation time from 120 to 108 h. Transcriptome and biochemical analyses demonstrated that both antioxidant enzyme-mediated oxidative stress defense system, such as superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX), and nonenzymatic antioxidant system, such as glutathione, were enhanced in the presence of elastin peptide, suggesting elastin peptide relieving oxidative stresses is involved in many pathways. In order to further investigate the relationship between oxidative stress defense and malic acid productivity, we overexpressed three enzymes (Sod1, CAT, Tps1) related to oxidation resistance in A. niger, respectively, and these resulting strains display varying degree of improvement in malic acid productivity. Especially, the strain overexpressing the Sod1 gene achieved a malate titer of 91.85 ± 2.58 g/L in 96 h, corresponding to a productivity of 0.96 g/L/h, which performs better than elastin peptide addition. CONCLUSIONS Our investigation provides an excellent reference for alleviating the stress of the fungal fermentation process and improving fermentation efficiency.
Collapse
Affiliation(s)
- Na Wu
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China ,grid.260474.30000 0001 0089 5711College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Mingyan Xing
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China
| | - Yaru Chen
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China
| | - Chi Zhang
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China
| | - Yingfeng Li
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China ,grid.260474.30000 0001 0089 5711College of Life Sciences, Nanjing Normal University, Nanjing, 210046 China
| | - Ping Song
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China
| | - Qing Xu
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China
| | - Hao Liu
- grid.413109.e0000 0000 9735 6249Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science & Technology, Tianjin, 300457 China
| | - He Huang
- grid.260474.30000 0001 0089 5711School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023 China ,grid.412022.70000 0000 9389 5210College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, 211800 China
| |
Collapse
|
31
|
Wu N, Zhang J, Chen Y, Xu Q, Song P, Li Y, Li K, Liu H. Recent advances in microbial production of L-malic acid. Appl Microbiol Biotechnol 2022; 106:7973-7992. [PMID: 36370160 DOI: 10.1007/s00253-022-12260-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/19/2022] [Accepted: 10/23/2022] [Indexed: 11/14/2022]
Abstract
Over the last few decades, increasing concerns regarding fossil fuel depletion and excessive CO2 emissions have led to extensive fundamental studies and industrial trials regarding microbial chemical production. As an additive or precursor, L-malic acid has been shown to exhibit distinctive properties in the food, pharmaceutical, and daily chemical industries. L-malic acid is currently mainly fabricated through a fumarate hydratase-based biocatalytic conversion route, wherein petroleum-derived fumaric acid serves as a substrate. In this review, for the first time, we comprehensively describe the methods of malic acid strain transformation, raw material utilization, malic acid separation, etc., especially recent progress and remaining challenges for industrial applications. First, we summarize the various pathways involved in L-malic acid biosynthesis using different microorganisms. We also discuss several strain engineering strategies for improving the titer, yield, and productivity of L-malic acid. We illustrate the currently available alternatives for reducing production costs and the existing strategies for optimizing the fermentation process. Finally, we summarize the present challenges and future perspectives regarding the development of microbial L-malic acid production. KEY POINTS: • A range of wild-type, mutant, laboratory-evolved, and metabolically engineered strains which could produce L-malic acid were comprehensively described. • Alternative raw materials for reducing production costs and the existing strategies for optimizing the fermentation were sufficiently summarized. • The present challenges and future perspectives regarding the development of microbial L-malic acid production were elaboratively discussed.
Collapse
Affiliation(s)
- Na Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Jiahui Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Yaru Chen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Ping Song
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Yingfeng Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Ke Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China.
| | - Hao Liu
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China.
- Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science & Technology, Tianjin, China.
| |
Collapse
|
32
|
Kövilein A, Aschmann V, Zadravec L, Ochsenreither K. Optimization of l-malic acid production from acetate with Aspergillus oryzae DSM 1863 using a pH-coupled feeding strategy. Microb Cell Fact 2022; 21:242. [DOI: 10.1186/s12934-022-01961-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022] Open
Abstract
Abstract
Background
Malic acid, a dicarboxylic acid mainly used in the food industry, is currently produced from fossil resources. The utilization of low-cost substrates derived from biomass could render microbial processes economic. Such feedstocks, like lignocellulosic hydrolysates or condensates of fast pyrolysis, can contain high concentrations of acetic acid. Acetate is a suitable substrate for l-malic acid production with the filamentous fungus Aspergillus oryzae DSM 1863, but concentrations obtained so far are low. An advantage of this carbon source is that it can be used for pH control and simultaneous substrate supply in the form of acetic acid. In this study, we therefore aimed to enhance l-malate production from acetate with A. oryzae by applying a pH-coupled feeding strategy.
Results
In 2.5-L bioreactor fermentations, several feeding strategies were evaluated. Using a pH-coupled feed consisting of 10 M acetic acid, the malic acid concentration was increased about 5.3-fold compared to the batch process without pH control, resulting in a maximum titer of 29.53 ± 1.82 g/L after 264 h. However, it was not possible to keep both the pH and the substrate concentration constant during this fermentation. By using 10 M acetic acid set to a pH of 4.5, or with the repeated addition of NaOH, the substrate concentration could be maintained within a constant range, but these strategies did not prove beneficial as lower maximum titers and yields were obtained. Since cessation of malic acid production was observed in later fermentation stages despite carbon availability, a possible product inhibition was evaluated in shake flask cultivations. In these experiments, malate and succinate, which is a major by-product during malic acid production, were added at concentrations of up to 50 g/L, and it was found that A. oryzae is capable of organic acid production even at high product concentrations.
Conclusions
This study demonstrates that a suitable feeding strategy is necessary for efficient malic acid production from acetate. It illustrates the potential of acetate as carbon source for microbial production of the organic acid and provides useful insights which can serve as basis for further optimization.
Collapse
|
33
|
Kövilein A, Zadravec L, Hohmann S, Umpfenbach J, Ochsenreither K. Effect of process mode, nitrogen source and temperature on L-malic acid production with Aspergillus oryzae DSM 1863 using acetate as carbon source. Front Bioeng Biotechnol 2022; 10:1033777. [PMID: 36312560 PMCID: PMC9614319 DOI: 10.3389/fbioe.2022.1033777] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/04/2022] [Indexed: 11/21/2022] Open
Abstract
Malic acid, mainly used as acidulant and taste enhancer in the food industry, is currently produced from fossil resources. In this study, microbial L-malate production with the filamentous fungus A. oryzae using the carbon source acetate was evaluated. Acetate is for example contained in biomass-derived substrates such as lignocellulosic hydrolysates and condensates of fast pyrolysis, thus avoiding competition with food production. Since research on malic acid synthesis from acetate is limited and reported productivities and yields are low, this work aimed to improve the process. First, different cultivation temperatures were tested. This parameter was found to affect the ratio between malic and succinic acid, which is the major by-product of organic acid production with A. oryzae. At 32°C, the malate share was highest (53.7 ± 1.6%), while it was lowest at 38°C (43.3 ± 1.1%) whereas succinate represented the main product (51.5 ± 1.0%). Besides the temperature, the type of nitrogen source was also found to affect malate synthesis as well as biomass production. In the pre-culture, the biomass concentration was increased by a factor of 3.4–3.9, and germination started earlier with the complex nitrogen sources yeast extract, casein hydrolysate and peptone compared to the defined nitrogen source (NH4)2SO4. Especially with yeast extract, malate synthesis in the main culture was accelerated and the titer obtained after 48 h was about 2.6 times higher than that quantified with (NH4)2SO4. To reduce substrate inhibition in acetate medium, fed-batch and repeated-batch processes were evaluated using (NH4)2SO4 or yeast extract as nitrogen source. In the fed-batch process, the period of malate production was extended, and the maximum product concentration was increased to 11.49 ± 1.84 g/L with (NH4)2SO4 and 12.08 ± 1.25 g/L with yeast extract. In the repeated-batch process, the total acid production was highest within the first 240 h of fermentation, but optimization is required to maintain high production rates in later cycles. The lessons learned in this study will help in the development of further process strategies to maximize malate production using acetate as alternative substrate to the commonly used glucose.
Collapse
|
34
|
Borin GP, Oliveira JVDC. Assessing the intracellular primary metabolic profile of Trichoderma reesei and Aspergillus niger grown on different carbon sources. FRONTIERS IN FUNGAL BIOLOGY 2022; 3:998361. [PMID: 37746225 PMCID: PMC10512294 DOI: 10.3389/ffunb.2022.998361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 08/29/2022] [Indexed: 09/26/2023]
Abstract
Trichoderma reesei and Aspergillus niger are efficient biological platforms for the production of various industrial products, including cellulases and organic acids. Nevertheless, despite the extensive research on these fungi, integrated analyses of omics-driven approaches are still missing. In this study, the intracellular metabolic profile of T. reesei RUT-C30 and A. niger N402 strains grown on glucose, lactose, carboxymethylcellulose (CMC), and steam-exploded sugarcane bagasse (SEB) as carbon sources for 48 h was analysed by proton nuclear magnetic resonance. The aim was to verify the changes in the primary metabolism triggered by these substrates and use transcriptomics data from the literature to better understand the dynamics of the observed alterations. Glucose and CMC induced higher fungal growth whereas fungi grown on lactose showed the lowest dry weight. Metabolic profile analysis revealed that mannitol, trehalose, glutamate, glutamine, and alanine were the most abundant metabolites in both fungi regardless of the carbon source. These metabolites are of particular interest for the mobilization of carbon and nitrogen, and stress tolerance inside the cell. Their concomitant presence indicates conserved mechanisms adopted by both fungi to assimilate carbon sources of different levels of recalcitrance. Moreover, the higher levels of galactose intermediates in T. reesei suggest its better adaptation in lactose, whereas glycolate and malate in CMC might indicate activation of the glyoxylate shunt. Glycerol and 4-aminobutyrate accumulated in A. niger grown on CMC and lactose, suggesting their relevant role in these carbon sources. In SEB, a lower quantity and diversity of metabolites were identified compared to the other carbon sources, and the metabolic changes and higher xylanase and pNPGase activities indicated a better utilization of bagasse by A. niger. Transcriptomic analysis supported the observed metabolic changes and pathways identified in this work. Taken together, we have advanced the knowledge about how fungal primary metabolism is affected by different carbon sources, and have drawn attention to metabolites still unexplored. These findings might ultimately be considered for developing more robust and efficient microbial factories.
Collapse
Affiliation(s)
- Gustavo Pagotto Borin
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), São Paulo, Brazil
- Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| | - Juliana Velasco de Castro Oliveira
- Brazilian Biorenewables National Laboratory (LNBR), Brazilian Center for Research in Energy and Materials (CNPEM), São Paulo, Brazil
- Graduate Program in Genetics and Molecular Biology, Institute of Biology, University of Campinas (UNICAMP), São Paulo, Brazil
| |
Collapse
|
35
|
Yadav M, Sehrawat N, Kumar S, Sharma AK, Singh M, Kumar A. Malic acid: fermentative production and applications. PHYSICAL SCIENCES REVIEWS 2022. [DOI: 10.1515/psr-2022-0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
Microbial metabolites have gained lot of industrial interest. These are currently employed in various industries including pharmaceuticals, chemical, textiles, food etc. Organic acids are among the important microbial products. Production of microbial organic acids present numerous advantages like agro-industrial waste may be utilized as substrate, low production cost, natural in origin and production is environment friendly. Malic acid is an organic acid (C4 dicarboxylic acid) that can be produced by microbes. It is also useful in industrial sectors as food, chemicals, and pharmaceuticals etc. Production/extraction of malic acid has been reported from fruits, egg shells, microbes, via chemical synthesis, bio-transformation and from renewable sources. Microbial production of malic acid seems very promising due to various advantages and the approach is environment-friendly. In recent years, researchers have focused on fermentative microbial production of malic acid and possibility of using agro-industrial waste as raw substrates. In current article, malic acid production along with applications has been discussed with recent advances in the area.
Collapse
Affiliation(s)
- Mukesh Yadav
- Department of Biotechnology , Maharishi Markandeshwar (Deemed to be University) , Mullana-Ambala , India
| | - Nirmala Sehrawat
- Department of Biotechnology , Maharishi Markandeshwar (Deemed to be University) , Mullana-Ambala , India
| | - Sunil Kumar
- Department of Microbiology, Faculty of Bio-Medical Sciences , Kampala International University , Kampala , Uganda
| | - Anil Kumar Sharma
- Department of Biotechnology , Maharishi Markandeshwar (Deemed to be University) , Mullana-Ambala , India
| | - Manoj Singh
- Department of Biotechnology , Maharishi Markandeshwar (Deemed to be University) , Mullana-Ambala , India
| | - Amit Kumar
- Department of Biotechnology, School of Engineering and Technology , Sharda University , Greater Noida , U.P. , India
| |
Collapse
|
36
|
Cao W, Zhang L, Wu L, Zhang M, Liu J, Xie Z, Liu H. Identification and genetic characterization of mitochondrial citrate transporters in Aspergillus niger. Front Microbiol 2022; 13:1009491. [PMID: 36177470 PMCID: PMC9512666 DOI: 10.3389/fmicb.2022.1009491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Aspergillus niger is a major cell factory for citric acid production, and the process of citrate export from mitochondria to cytoplasm is predicted to be one of rate-limiting steps in citric acid accumulation. Currently, the mitochondrial citrate transporters (Ctps) in A. niger are not fully characterized. Here, six putative Ctp encoding genes (ctpA to ctpF) were identified based on their homology with a mitochondrial citrate transporter ScCtp1 from Saccharomyces cerevisiae. Disruption of individual ctpA to ctpF caused varying degrees of decline in citric acid accumulation at different fermentation stages, whereas a mutant strain S1696 with disruption of all six ctps showed complete loss of citiric acid production. S1696 also exhibited delayed growth, reduced conidia formation, and decreased pigmentogenesis. Exogenous addition of citrate partially restored the conidia formation and pigmentogenesis in S1696 mutant. Reintroduction of individual ctps (ctpA to ctpF) into S1696 at the amyA locus showed that ctpA, ctpB, and ctpD restored the citric acid titers to 88.5, 93.8, and 94.6% of the parent strain, respectively. Additionally, the formation of conidia and pigment production was partially restored after reintroduction of ctpA, ctpB, or ctpD. Overexpression of respective ctpA, ctpB, and ctpD in the parent strain resulted in increases in citric acid accumulation by 32.8, 19.3, and 24.2%, respectively. These results demonstrate that CtpA, CtpB, and CtpD play important roles in citric acid transport across the mitochondrial membrane and function in a redundant manner. Enhancement of citric acid transport process can serve as a target for boosting citric acid accumulation in A. niger.
Collapse
Affiliation(s)
- Wei Cao
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science and Technology, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
| | - Licheng Zhang
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Liu Wu
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Mingyi Zhang
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Jiao Liu
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science and Technology, Tianjin, China
| | - Zhoujie Xie
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science and Technology, Tianjin, China
| | - Hao Liu
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science and Technology, Tianjin, China
- National Technology Innovation Center of Synthetic Biology, Tianjin, China
- *Correspondence: Hao Liu,
| |
Collapse
|
37
|
Sreekala AGV, Ismail MHB, Nathan VK. Biotechnological interventions in food waste treatment for obtaining value-added compounds to combat pollution. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62755-62784. [PMID: 35802320 DOI: 10.1007/s11356-022-21794-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 06/28/2022] [Indexed: 06/15/2023]
Abstract
Over the last few decades, the globe is facing tremendous effects due to the unnecessary piling of municipal solid waste among which food waste holds a greater portion. This practice not only affects the environment in terms of generating greenhouse gas emissions but when left dumped in landfills will also trigger poverty and malnutrition. This review focuses on the global trend in food waste management strategies involved in the effective utilization of food waste to produce various value-added products in a microbiology aspect, thereby diminishing the negative impacts caused by the unnecessary side effects of non-renewable energy sources. The review also detailed the efficiency of microorganisms in the production of various bio-energies as well. Further, recent attempts to the exploitation of genetically modified microorganisms in producing value-added products were enlisted. This also attempted to address food waste valorization techniques, the combined applications of various processes for an enhanced yield of different compounds, and addressed various challenges. Further, the current challenges involved in various processes and the effective measures to tackle them in the future have been addressed. Thus, the present review has successfully addressed the circular bio-economy in food waste valorization.
Collapse
Affiliation(s)
| | - Muhammad Heikal Bin Ismail
- Department of Chemical and Environmental Engineering, Faculty of Engineering, Universiti Putra, Putrajaya, Malaysia
| | - Vinod Kumar Nathan
- School of Chemical and Biotechnology, SASTRA Deemed to Be University, Thanjavur, 613 401, Tamil Nadu, India.
| |
Collapse
|
38
|
Wang Y, Guo Y, Cao W, Liu H. Synergistic effects on itaconic acid production in engineered Aspergillus niger expressing the two distinct biosynthesis clusters from Aspergillus terreus and Ustilago maydis. Microb Cell Fact 2022; 21:158. [PMID: 35953829 PMCID: PMC9367143 DOI: 10.1186/s12934-022-01881-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Itaconic acid (IA) is a versatile platform chemical widely used for the synthesis of various polymers and current methods for IA production based on Aspergillus terreus fermentation are limited in terms of process efficiency and productivity. To construct more efficient IA production strains, A. niger was used as a chassis for engineering IA production by assembling the key components of IA biosynthesis pathways from both A. terreus and Ustilago maydis. RESULTS Recombinant A. niger S1596 overexpressing the A. terreus IA biosynthesis genes cadA, mttA, mfsA produced IA of 4.32 g/L, while A. niger S2120 overexpressing the U. maydis IA gene cluster adi1, tad1, mtt1, itp1 achieved IA of 3.02 g/L. Integration of the two IA production pathways led to the construction of A. niger S2083 with IA titers of 5.58 g/L. Increasing cadA copy number in strain S2083 created strain S2209 with titers of 7.99 g/L and deleting ictA to block IA degradation in S2209 created strain S2288 with IA titers of 8.70 g/L. Overexpressing acoA to enhance the supply of IA precursor in strain S2288 generated strain S2444 with IA titers of 9.08 g/L in shake flask. CONCLUSION Recombinant A. niger overexpressing the U. maydis IA biosynthesis pathway was capable of IA accumulation. Combined expression of the two IA biosynthesis pathways from A. terreus and U. maydis in A. niger resulted in much higher IA titers. Furthermore, increasing cadA copy number, deleting ictA to block IA degradation and overexpressing acoA to enhance IA precursor supply all showed beneficial effects on IA accumulation.
Collapse
Affiliation(s)
- Yaqi Wang
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, People's Republic of China
| | - Yufei Guo
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, People's Republic of China
| | - Wei Cao
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, People's Republic of China.,Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science & Technology, Tianjin, 300457, People's Republic of China.,National Technology Innovation Center of Synthetic Biology, Tianjin, 300308, People's Republic of China
| | - Hao Liu
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, 300457, People's Republic of China. .,Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science & Technology, Tianjin, 300457, People's Republic of China. .,National Technology Innovation Center of Synthetic Biology, Tianjin, 300308, People's Republic of China.
| |
Collapse
|
39
|
Zhang J, Wu N, Ou W, Li Y, Liang Y, Peng C, Li Y, Xu Q, Tong Y. Peptide supplementation relieves stress and enhances glycolytic flux in filamentous fungi during organic acid bioproduction. Biotechnol Bioeng 2022; 119:2471-2481. [PMID: 35665482 DOI: 10.1002/bit.28152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 05/09/2022] [Accepted: 05/24/2022] [Indexed: 11/07/2022]
Abstract
Filamentous fungi occupy a uniquely favorable position in the bioproduction of organic acids. Intracellular stress is the main stimulator in filamentous fungi to produce and accumulate organic acids with high flux. However, stress can affect the physiological activities of filamentous fungi, thereby deteriorating their fermentation performance. Herein, we report that peptide supplementation during Rhizopus oryzae fermentation significantly improved fumaric acid production. Specifically, fumaric acid productivity was elevated by approximately 100%, fermentation duration was shortened from 72 to 36 h, while maintaining the final titer. Furthermore, transcriptome profile analysis and biochemical assays indicated that the overall capabilities of the stress defense systems (enzymatic and nonenzymatic) were significantly improved in R. oryzae. Consequently, glycolytic metabolism was distinctly enhanced, which eventually resulted in improved fumaric acid production and reduced fermentation duration. We expect our findings and efforts to provide essential insights into the optimization of the fermentation performance of filamentous fungi in industrial biotechnology and fermentation engineering.
Collapse
Affiliation(s)
- Jiahui Zhang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Na Wu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Wen Ou
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Yingfeng Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Yingchao Liang
- National Engineering Research Center of Corn Deep Processing, Jilin COFCO Biochemistry Co., Ltd., Changchun, China
| | - Chao Peng
- Nutrition & Health Research Institute, COFCO Corporation, Beijing, China
| | - Yi Li
- National Engineering Research Center of Corn Deep Processing, Jilin COFCO Biochemistry Co., Ltd., Changchun, China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Yi Tong
- National Engineering Research Center of Corn Deep Processing, Jilin COFCO Biochemistry Co., Ltd., Changchun, China.,Nutrition & Health Research Institute, COFCO Corporation, Beijing, China
| |
Collapse
|
40
|
Enzymatic Preparation of l-Malate in a Reaction System with Product Separation and Enzyme Recycling. Catalysts 2022. [DOI: 10.3390/catal12060587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Reaction coupling separation systems using calcium fumarate as a substrate can break the reaction equilibrium and promote the production of l-malate. However, the low reusability and stability of fumarase limit its further application. In this study, partially purified fumarase of Thermus thermophilus (87.0 U mg−1) was immobilized within konjac-κ-carrageenan beads. An amalgamation of konjac and carrageenan gum (2%) was used to form the beads, and polyethylene polyamine (0.2%) and glutaraldehyde (0.1%) were used as the cross-linking agents. The pH and temperature profiles of free and immobilized fumarases were remarkably similar. The diffusion limit of immobilized fumarase caused a decline in the maximal velocity (Vmax), whereas the kinetic constant (Km) value increased. Optimization of the parameters for biotransformation by immobilized fumarase showed that 88.3% conversion of 200 mM calcium fumarate could be achieved at 55 °C within 8 h. The beads were stored for 30 days at 4 °C with minimal loss in activity and were reusable for up to 20 cycles with 78.1% relative activity. By recycling the reaction supernatant, a total amount of 3.98 M calcium fumarate was obtained with a conversion of 99.5%, which is the highest value ever reported.
Collapse
|
41
|
Evaluation of Aspergillus niger Six Constitutive Strong Promoters by Fluorescent-Auxotrophic Selection Coupled with Flow Cytometry: A Case for Citric Acid Production. J Fungi (Basel) 2022; 8:jof8060568. [PMID: 35736051 PMCID: PMC9224621 DOI: 10.3390/jof8060568] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/19/2022] [Accepted: 05/24/2022] [Indexed: 02/05/2023] Open
Abstract
Aspergillus niger is an important industrial workhorse for the biomanufacturing of organic acids, proteins, etc. Well-controlled genetic regulatory elements, including promoters, are vital for strain engineering, but available strong promoters for A. niger are limited. Herein, to efficiently assess promoters, we developed an accurate and intuitive fluorescent-auxotrophic selection workflow based on mCherry, pyrG, CRISPR/Cas9 system, and flow cytometry. With this workflow, we characterized six endogenous constitutive promoters in A. niger. The endogenous glyceraldehyde-3-phosphate dehydrogenase promoter PgpdAg showed a 2.28-fold increase in promoter activity compared with the most frequently used strong promoter PgpdAd from A. nidulans. Six predicted conserved motifs, including the gpdA-box, were verified to be essential for the PgpdAg activity. To demonstrate its application, the promoter PgpdAg was used for enhancing the expression of citrate exporter cexA in a citric acid-producing isolate D353.8. Compared with the cexA controlled by PgpdAd, the transcription level of the cexA gene driven by PgpdAg increased by 2.19-fold, which is consistent with the promoter activity assessment. Moreover, following cexA overexpression, several genes involved in carbohydrate transport and metabolism were synergically upregulated, resulting in up to a 2.48-fold increase in citric acid titer compared with that of the parent strain. This study provides an intuitive workflow to speed up the quantitative evaluation of A. niger promoters and strong constitutive promoters for fungal cell factory construction and strain engineering.
Collapse
|
42
|
Valorization of a Pyrolytic Aqueous Condensate and Its Main Components for L-Malic Acid Production with Aspergillus oryzae DSM 1863. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8030107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pyrolytic aqueous condensate (PAC) might serve as a cost-effective substrate for microbial malic acid production, as it is an unused side stream of the fast pyrolysis of lignocellulosic biomass that contains acetol and acetate as potential carbon sources. In the present study, shake flask cultures were performed to evaluate the suitability of acetol and its combination with acetate as substrates for growth and L-malate production with the filamentous fungus Aspergillus oryzae. Acetol concentrations of up to 40 g/L were shown to be utilized for fungal growth. In combination with acetate, co-metabolization of both substrates for biomass and malate formation was observed, although the maximum tolerated acetol concentration decreased to 20 g/L. Furthermore, malate production on PAC detoxified by a combination of rotary evaporation, overliming and activated carbon treatment was studied. In shake flasks, cultivation using 100% PAC resulted in the production of 3.37 ± 0.61 g/L malate, which was considerably improved by pH adjustment up to 9.77 ± 0.55 g/L. A successful scale-up to 0.5-L bioreactors was conducted, achieving comparable yields and productivities to the shake flask cultures. Accordingly, fungal malate production using PAC was successfully demonstrated, paving the way for a bio-based production of the acid.
Collapse
|
43
|
Liu J, Liu J, Guo L, Liu J, Chen X, Liu L, Gao C. Advances in microbial synthesis of bioplastic monomers. ADVANCES IN APPLIED MICROBIOLOGY 2022; 119:35-81. [DOI: 10.1016/bs.aambs.2022.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
44
|
Madhavan A, Arun KB, Sindhu R, Alphonsa Jose A, Pugazhendhi A, Binod P, Sirohi R, Reshmy R, Kumar Awasthi M. Engineering interventions in industrial filamentous fungal cell factories for biomass valorization. BIORESOURCE TECHNOLOGY 2022; 344:126209. [PMID: 34715339 DOI: 10.1016/j.biortech.2021.126209] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 05/15/2023]
Abstract
Filamentous fungi possess versatile capabilities for synthesizing a variety of valuable bio compounds, including enzymes, organic acids and small molecule secondary metabolites. The advancements of genetic and metabolic engineering techniques and the availability of sequenced genomes discovered their potential as expression hosts for recombinant protein production. Remarkably, plant-biomass degrading filamentous fungi show the unique capability to decompose lignocellulose, an extremely recalcitrant biopolymer. The basic biochemical approaches have motivated several industrial processes for lignocellulose biomass valorisation into fermentable sugars and other biochemical for biofuels, biomolecules, and biomaterials. The review gives insight into current trends in engineering filamentous fungi for enzymes, fuels, and chemicals from lignocellulose biomass. This review describes the variety of enzymes and compounds that filamentous fungi produce, engineering of filamentous fungi for biomass valorisation with a special focus on lignocellulolytic enzymes and other bulk chemicals.
Collapse
Affiliation(s)
- Aravind Madhavan
- Rajiv Gandhi Centre for Biotechnology, Jagathy, Trivandrum 695 014, India.
| | - K B Arun
- Rajiv Gandhi Centre for Biotechnology, Jagathy, Trivandrum 695 014, India
| | - Raveendran Sindhu
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Trivandrum 695 019, India
| | - Anju Alphonsa Jose
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Trivandrum 695 019, India
| | | | - Parameswaran Binod
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology, Trivandrum 695 019, India
| | - Ranjna Sirohi
- Department of Chemical & Biological Engineering, Korea University, Seoul 136713, Republic of Korea; Centre for Energy & Environmental Sustainability, Lucknow 226001. Uttar Pradesh, India
| | - R Reshmy
- Post Graduate and Research Department of Chemistry, Bishop Moore College, Mavelikara 690 110, Kerala, India
| | - Mukesh Kumar Awasthi
- College of Natural Resources and Environment, Northwest A & F University, Yangling, Shaanxi 712 100, PR China
| |
Collapse
|
45
|
Current Progress in Production of Building-Block Organic Acids by Consolidated Bioprocessing of Lignocellulose. FERMENTATION-BASEL 2021. [DOI: 10.3390/fermentation7040248] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Several organic acids have been indicated among the top value chemicals from biomass. Lignocellulose is among the most attractive feedstocks for biorefining processes owing to its high abundance and low cost. However, its highly complex nature and recalcitrance to biodegradation hinder development of cost-competitive fermentation processes. Here, current progress in development of single-pot fermentation (i.e., consolidated bioprocessing, CBP) of lignocellulosic biomass to high value organic acids will be examined, based on the potential of this approach to dramatically reduce process costs. Different strategies for CBP development will be considered such as: (i) design of microbial consortia consisting of (hemi)cellulolytic and valuable-compound producing strains; (ii) engineering of microorganisms that combine biomass-degrading and high-value compound-producing properties in a single strain. The present review will mainly focus on production of organic acids with application as building block chemicals (e.g., adipic, cis,cis-muconic, fumaric, itaconic, lactic, malic, and succinic acid) since polymer synthesis constitutes the largest sector in the chemical industry. Current research advances will be illustrated together with challenges and perspectives for future investigations. In addition, attention will be dedicated to development of acid tolerant microorganisms, an essential feature for improving titer and productivity of fermentative production of acids.
Collapse
|
46
|
Wei Z, Xu Y, Xu Q, Cao W, Huang H, Liu H. Microbial Biosynthesis of L-Malic Acid and Related Metabolic Engineering Strategies: Advances and Prospects. Front Bioeng Biotechnol 2021; 9:765685. [PMID: 34660563 PMCID: PMC8511312 DOI: 10.3389/fbioe.2021.765685] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Malic acid, a four-carbon dicarboxylic acid, is widely used in the food, chemical and medical industries. As an intermediate of the TCA cycle, malic acid is one of the most promising building block chemicals that can be produced from renewable sources. To date, chemical synthesis or enzymatic conversion of petrochemical feedstocks are still the dominant mode for malic acid production. However, with increasing concerns surrounding environmental issues in recent years, microbial fermentation for the production of L-malic acid was extensively explored as an eco-friendly production process. The rapid development of genetic engineering has resulted in some promising strains suitable for large-scale bio-based production of malic acid. This review offers a comprehensive overview of the most recent developments, including a spectrum of wild-type, mutant, laboratory-evolved and metabolically engineered microorganisms for malic acid production. The technological progress in the fermentative production of malic acid is presented. Metabolic engineering strategies for malic acid production in various microorganisms are particularly reviewed. Biosynthetic pathways, transport of malic acid, elimination of byproducts and enhancement of metabolic fluxes are discussed and compared as strategies for improving malic acid production, thus providing insights into the current state of malic acid production, as well as further research directions for more efficient and economical microbial malic acid production.
Collapse
Affiliation(s)
- Zhen Wei
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Yongxue Xu
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China
| | - Qing Xu
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Wei Cao
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China.,Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science & Technology, Tianjin, China
| | - He Huang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Hao Liu
- MOE Key Laboratory of Industrial Fermentation Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin, China.,Tianjin Engineering Research Center of Microbial Metabolism and Fermentation Process Control, Tianjin University of Science & Technology, Tianjin, China
| |
Collapse
|
47
|
Something old, something new: challenges and developments in Aspergillus niger biotechnology. Essays Biochem 2021; 65:213-224. [PMID: 33955461 PMCID: PMC8314004 DOI: 10.1042/ebc20200139] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022]
Abstract
The filamentous ascomycete fungus Aspergillus niger is a prolific secretor of organic acids, proteins, enzymes and secondary metabolites. Throughout the last century, biotechnologists have developed A. niger into a multipurpose cell factory with a product portfolio worth billions of dollars each year. Recent technological advances, from genome editing to other molecular and omics tools, promise to revolutionize our understanding of A. niger biology, ultimately to increase efficiency of existing industrial applications or even to make entirely new products. However, various challenges to this biotechnological vision, many several decades old, still limit applications of this fungus. These include an inability to tightly control A. niger growth for optimal productivity, and a lack of high-throughput cultivation conditions for mutant screening. In this mini-review, we summarize the current state-of-the-art for A. niger biotechnology with special focus on organic acids (citric acid, malic acid, gluconic acid and itaconic acid), secreted proteins and secondary metabolites, and discuss how new technological developments can be applied to comprehensively address a variety of old and persistent challenges.
Collapse
|
48
|
Cairns TC, Zheng X, Zheng P, Sun J, Meyer V. Turning Inside Out: Filamentous Fungal Secretion and Its Applications in Biotechnology, Agriculture, and the Clinic. J Fungi (Basel) 2021; 7:535. [PMID: 34356914 PMCID: PMC8307877 DOI: 10.3390/jof7070535] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/14/2021] [Accepted: 06/25/2021] [Indexed: 12/15/2022] Open
Abstract
Filamentous fungi are found in virtually every marine and terrestrial habitat. Vital to this success is their ability to secrete a diverse range of molecules, including hydrolytic enzymes, organic acids, and small molecular weight natural products. Industrial biotechnologists have successfully harnessed and re-engineered the secretory capacity of dozens of filamentous fungal species to make a diverse portfolio of useful molecules. The study of fungal secretion outside fermenters, e.g., during host infection or in mixed microbial communities, has also led to the development of novel and emerging technological breakthroughs, ranging from ultra-sensitive biosensors of fungal disease to the efficient bioremediation of polluted environments. In this review, we consider filamentous fungal secretion across multiple disciplinary boundaries (e.g., white, green, and red biotechnology) and product classes (protein, organic acid, and secondary metabolite). We summarize the mechanistic understanding for how various molecules are secreted and present numerous applications for extracellular products. Additionally, we discuss how the control of secretory pathways and the polar growth of filamentous hyphae can be utilized in diverse settings, including industrial biotechnology, agriculture, and the clinic.
Collapse
Affiliation(s)
- Timothy C. Cairns
- Chair of Applied and Molecular Microbiology, Institute of Biotechnology, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany
| | - Xiaomei Zheng
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; (X.Z.); (P.Z.); (J.S.)
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Ping Zheng
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; (X.Z.); (P.Z.); (J.S.)
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Jibin Sun
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China; (X.Z.); (P.Z.); (J.S.)
- Key Laboratory of Systems Microbial Biotechnology, Chinese Academy of Sciences, Tianjin 300308, China
| | - Vera Meyer
- Chair of Applied and Molecular Microbiology, Institute of Biotechnology, Technische Universität Berlin, Straße des 17. Juni 135, 10623 Berlin, Germany
| |
Collapse
|
49
|
Karaffa L, Fekete E, Kubicek CP. The Role of Metal Ions in Fungal Organic Acid Accumulation. Microorganisms 2021; 9:1267. [PMID: 34200938 PMCID: PMC8230503 DOI: 10.3390/microorganisms9061267] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 11/22/2022] Open
Abstract
Organic acid accumulation is probably the best-known example of primary metabolic overflow. Both bacteria and fungi are capable of producing various organic acids in large amounts under certain conditions, but in terms of productivity-and consequently, of commercial importance-fungal platforms are unparalleled. For high product yield, chemical composition of the growth medium is crucial in providing the necessary conditions, of which the concentrations of four of the first-row transition metal elements, manganese (Mn2+), iron (Fe2+), copper (Cu2+) and zinc (Zn2+) stand out. In this paper we critically review the biological roles of these ions, the possible biochemical and physiological consequences of their influence on the accumulation of the most important mono-, di- and tricarboxylic as well as sugar acids by fungi, and the metal ion-related aspects of submerged organic acid fermentations, including the necessary instrumental analytics. Since producing conditions are associated with a cell physiology that differs strongly to what is observed under "standard" growth conditions, here we consider papers and patents only in which organic acid accumulation levels achieved at least 60% of the theoretical maximum yield, and the actual trace metal ion concentrations were verified.
Collapse
Affiliation(s)
- Levente Karaffa
- Department of Biochemical Engineering, Faculty of Science and Technology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Erzsébet Fekete
- Department of Biochemical Engineering, Faculty of Science and Technology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Christian P. Kubicek
- Institute of Chemical, Environmental & Bioscience Engineering, TU Wien, A-1060 Vienna, Austria;
| |
Collapse
|
50
|
Ji L, Wang J, Luo Q, Ding Q, Tang W, Chen X, Liu L. Enhancing L-malate production of Aspergillus oryzae by nitrogen regulation strategy. Appl Microbiol Biotechnol 2021; 105:3101-3113. [PMID: 33818672 DOI: 10.1007/s00253-021-11149-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/14/2021] [Accepted: 01/26/2021] [Indexed: 12/01/2022]
Abstract
Regulating morphology engineering and fermentation of Aspergillus oryzae makes it possible to increase the titer of L-malate. However, the existing L-malate-producing strain has limited L-malate production capacity and the fermentation process is insufficiently mature, which cannot meet the needs of industrial L-malate production. To further increase the L-malate production capacity of A. oryzae, we screened out a mutant strain (FMME-S-38) that produced 79.8 g/L L-malate in 250-mL shake flasks, using a newly developed screening system based on colony morphology on the plate. We further compared the extracellular nitrogen (N1) and intracellular nitrogen (N2) contents of the control and mutant strain (FMME-S-38) to determine the relationship between the curve of nitrogen content (N1 and N2) and the L-malate titer. This correlation was then used to optimize the conditions for developing a novel nitrogen supply strategy (initial tryptone concentration of 6.5 g/L and feeding with 3 g/L tryptone at 24 h). Fermentation in a 7.5-L fermentor under the optimized conditions further increased the titer and productivity of L-malate to 143.3 g/L and 1.19 g/L/h, respectively, corresponding to 164.9 g/L and 1.14 g/L/h in a 30-L fermentor. This nitrogen regulation-based strategy cannot only enhance industrial-scale L-malate production but also has generalizability and the potential to increase the production of similar metabolites.Key Points• Construction of a new screening system based on colony morphology on the plate.• A novel nitrogen regulation strategy used to regulate the production of L-malate.• A nitrogen supply strategy used to maximize the production of L-malate.
Collapse
Affiliation(s)
- Lihao Ji
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Ju Wang
- College of Food Engineering, Anhui Science and Technology University, Chuzhou, 233100, Anhui, China
| | - Qiuling Luo
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Qiang Ding
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Wenxiu Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Xiulai Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China.,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China
| | - Liming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, 214122, China. .,International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|